Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

AMPK modulates Hippo pathway activity to regulate energy homeostasis

Abstract

The Hippo pathway was discovered as a conserved tumour suppressor pathway restricting cell proliferation and apoptosis. However, the upstream signals that regulate the Hippo pathway in the context of organ size control and cancer prevention are largely unknown. Here, we report that glucose, the ubiquitous energy source used for ATP generation, regulates the Hippo pathway downstream effector YAP. We show that both the Hippo pathway and AMP-activated protein kinase (AMPK) were activated during glucose starvation, resulting in phosphorylation of YAP and contributing to its inactivation. We also identified glucose-transporter 3 (GLUT3) as a YAP-regulated gene involved in glucose metabolism. Together, these results demonstrate that glucose-mediated energy homeostasis is an upstream event involved in regulation of the Hippo pathway and, potentially, an oncogenic function of YAP in promoting glycolysis, thereby providing an exciting link between glucose metabolism and the Hippo pathway in tissue maintenance and cancer prevention.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Glucose homeostasis controls YAP phosphorylation and localization.
Figure 2: Release from energy stress activates YAP.
Figure 3: AMPK associates with and phosphorylates YAP.
Figure 4: AMPK phosphorylates YAP at S61 and suppresses its transcriptional activity.
Figure 5: Energy stress increases LATS kinase activation independent of MST kinases.
Figure 6: YAP regulates GLUT3 transcription in the glucose metabolic pathway.
Figure 7: YAP and GLUT3 expression positively correlate with each other in human colon and liver cancers.

Similar content being viewed by others

References

  1. Pan, D. The hippo signaling pathway in development and cancer. Dev. Cell 19, 491–505 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zhao, B., Li, L., Lei, Q. & Guan, K. L. The Hippo-YAP pathway in organ size control and tumorigenesis: an updated version. Genes Dev. 24, 862–874 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Halder, G. & Johnson, R. L. Hippo signaling: growth control and beyond. Development 138, 9–22 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhao, B., Li, L., Tumaneng, K., Wang, C. Y. & Guan, K. L. A coordinated phosphorylation by Lats and CK1 regulates YAP stability through SCFβ−TRCP. Genes Dev. 24, 72–85 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Jiao, S. et al. A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer. Cancer Cell 25, 166–180 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Zhang, W. et al. VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex. Cell Res. 24, 331–343 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lu, L. et al. Hippo signaling is a potent in vivo growth and tumor suppressor pathway in the mammalian liver. Proc. Natl Acad. Sci. USA 107, 1437–1442 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cai, J. et al. The Hippo signaling pathway restricts the oncogenic potential of an intestinal regeneration program. Genes Dev. 24, 2383–2388 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lee, K. P. et al. The Hippo-Salvador pathway restrains hepatic oval cell proliferation, liver size, and liver tumorigenesis. Proc. Natl Acad. Sci. USA 107, 8248–8253 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Camargo, F. D. et al. YAP1 increases organ size and expands undifferentiated progenitor cells. Curr. Biol. 17, 2054–2060 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Dong, J. et al. Elucidation of a universal size-control mechanism in Drosophila and mammals. Cell 130, 1120–1133 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mo, J. S., Park, H. W. & Guan, K. L. The Hippo signaling pathway in stem cell biology and cancer. EMBO Rep. 15, 642–656 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhao, B. et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 21, 2747–2761 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhao, B. et al. Cell detachment activates the Hippo pathway via cytoskeleton reorganization to induce anoikis. Genes Dev. 26, 54–68 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Halder, G., Dupont, S. & Piccolo, S. Transduction of mechanical and cytoskeletal cues by YAP and TAZ. Nat. Rev. Mol. Cell Biol. 13, 591–600 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. Dupont, S. et al. Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183 (2011).

    Article  CAS  PubMed  Google Scholar 

  17. Yu, F. X. et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 150, 780–791 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Jones, R. G. & Thompson, C. B. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev. 23, 537–548 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cairns, R. A., Harris, I. S. & Mak, T. W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 11, 85–95 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Egan, D. F. et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331, 456–461 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Crute, B. E., Seefeld, K., Gamble, J., Kemp, B. E. & Witters, L. A. Functional domains of the alpha1 catalytic subunit of the AMP-activated protein kinase. J. Biol. Chem. 273, 35347–35354 (1998).

    Article  CAS  PubMed  Google Scholar 

  22. Hao, Y., Chun, A., Cheung, K., Rashidi, B. & Yang, X. Tumor suppressor LATS1 is a negative regulator of oncogene YAP. J. Biol. Chem. 283, 5496–5509 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Fujishiro, S. H. et al. ERK1/2 phosphorylate GEF-H1 to enhance its guanine nucleotide exchange activity toward RhoA. Biochem. Biophys. Res. Commun. 368, 162–167 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Wu, S. Z. et al. Akt and RhoA activation in response to high glucose require caveolin-1 phosphorylation in mesangial cells. Am. J. Physiol. Renal Physiol. 306, F1308–F1317 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Zhang, Y., Peng, F., Gao, B., Ingram, A. J. & Krepinsky, J. C. High glucose-induced RhoA activation requires caveolae and PKCbeta1-mediated ROS generation. Am. J. Physiol. Renal Physiol. 302, F159–F172 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Zhao, B. et al. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev. 22, 1962–1971 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Stubbs, M., McSheehy, P. M., Griffiths, J. R. & Bashford, C. L. Causes and consequences of tumour acidity and implications for treatment. Mol. Med. Today 6, 15–19 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Zhang, H., Pasolli, H. A. & Fuchs, E. Yes-associated protein (YAP) transcriptional coactivator functions in balancing growth and differentiation in skin. Proc. Natl Acad. Sci. USA 108, 2270–2275 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yamamoto, T. et al. Over-expression of facilitative glucose transporter genes in human cancer. Biochem. Biophys. Res. Commun. 170, 223–230 (1990).

    Article  CAS  PubMed  Google Scholar 

  30. Mellanen, P., Minn, H., Grenman, R. & Harkonen, P. Expression of glucose transporters in head-and-neck tumors. Int. J. Cancer 56, 622–629 (1994).

    Article  CAS  PubMed  Google Scholar 

  31. Boado, R. J., Black, K. L. & Pardridge, W. M. Gene expression of GLUT3 and GLUT1 glucose transporters in human brain tumors. Brain Res. Mol. Brain Res. 27, 51–57 (1994).

    Article  CAS  PubMed  Google Scholar 

  32. Younes, M., Brown, R. W., Stephenson, M., Gondo, M. & Cagle, P. T. Overexpression of Glut1 and Glut3 in stage I nonsmall cell lung carcinoma is associated with poor survival. Cancer 80, 1046–1051 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. Ayala, F. R. et al. GLUT1 and GLUT3 as potential prognostic markers for oral squamous cell carcinoma. Molecules 15, 2374–2387 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ha, T. K. & Chi, S. G. CAV1/caveolin 1 enhances aerobic glycolysis in colon cancer cells via activation of SLC2A3/GLUT3 transcription. Autophagy 8, 1684–1685 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Mo, J-S. et al. Cellular energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nat. Cell Biol. 17, http://dx.doi.org/10.1038/ncb3111 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. DeRan, M. et al. Energy stress regulates Hippo-YAP signaling involving AMPK-mediated regulation of angiomotin-like 1 protein. Cell Rep. 9, 495–503 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Shevchenko, A., Wilm, M., Vorm, O. & Mann, M. Mass spectrometric sequencing of proteins silver-stained polyacrylamide gels. Anal. Chem. 68, 850–858 (1996).

    Article  CAS  PubMed  Google Scholar 

  38. Peng, J. & Gygi, S. P. Proteomics: the move to mixtures. J. Mass Spectrom. 36, 1083–1091 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Eng, J. K., McCormack, A. L. & Yates, J. R. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J. Am. Soc. Mass Spectrom. 5, 976–989 (1994).

    Article  CAS  PubMed  Google Scholar 

  40. Beausoleil, S. A., Villen, J., Gerber, S. A., Rush, J. & Gygi, S. P. A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nat. Biotechnol. 24, 1285–1292 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all of our colleagues in the Chen laboratory for insightful discussion and technical assistance, especially J. Yuan, G. Ghosal and B. C. Nair. We also thank J-I. Park, L. Ma, Y. Sun, J. Zhang, A. Lin and H. Lee for reagents, insightful suggestions and comments on this work. We thank F-X. Yu (University of California, San Diego) for technical help. We thank the shRNA-ORFeome Core Facility at MD Anderson Cancer Center for the ORFs and shRNAs. We thank B. Viollet for providing AMPK wild-type and knockout MEFs. We thank K. Hale for proof-reading the manuscript. We thank R. Tomaino for assistance with the mass spectrometry analysis. This work was supported in part by the US Department of Defense Era of Hope research scholar award to J.C. (W81XWH-09-1-0409 and W81XWH-05-1-0470). This work was partly supported by the U.S. National Cancer Institute through the MD Anderson Cancer Center Support Grant (CA016672).

Author information

Authors and Affiliations

Authors

Contributions

W.W. performed all of the experiments with assistance from Z-D.X., X.L., K.E.A., B.G., R.L.J. and J.C. W.W. and J.C. designed the experiments. J.C. supervised the study. W.W. and J.C. wrote the manuscript. All authors commented on the manuscript.

Corresponding author

Correspondence to Junjie Chen.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 2 Identification of AMPK phosphorylation sites on YAP by mass spectrometry.

Bacteria-purified GST-YAP (2 μg) was used as substrate for AMPK in vitro kinase assay in the presence of cold ATP (500 μM) as described in the Supplementary Methods. Phosphorylation analysis of YAP was performed by the Taplin Mass Spectrometry Facility. Information on the identified phospho-peptides is shown.

Supplementary Figure 3 Validation of YAP S61 phospho-antibody.

(A) 2-DG induced YAP 5SA mutant phosphorylation. HEK293A cells were transfected with indicated plasmids and treated with glucose-free medium supplemented with 2-DG for 4 hours. Cell lysates were subjected to western blotting. (B) In vitro kinase assay using purified AMPK kinase. SFB-AMPKα1 or its kinase-dead mutant (K47R) was co-expressed with Flag-AMPKβ2 or Flag-AMPKγ2 plasmid in 293T cells for 48 hours. AMPK complex was purified by S protein beads and subjected to the in vitrokinase assay, where GST-ACC (residues 1-130) (2 μg) was purified from bacterial and taken as the substrate. Reaction samples were subjected to western blotting with indicated antibodies to verify the AMPK-dependent phosphorylation of ACC. (C–D) Validation of YAP S61 phospho-antibody. YAP S61 phospho-antibody was used to detect AMPK-mediated YAP phosphorylation by the in vitro kinase assay (C). Short exp and long exp represent short exposure and long exposure respectively. YAP S61 antibody was also used to detect AMPK active truncation mutant-mediated YAP phosphorylation in vivo (D). AMPK kinase-dead mutant (K47R) and active AMPK truncation dead mutant (T172A) were used as negative controls for AMPK. YAP-S61A mutant was used as the control for wild-type YAP. (E) Sequence alignment of the AMPK phosphorylation sites in YAP from different species and other known AMPK substrates. Uncropped images of western blots are shown in Supplementary Fig. 7.

Supplementary Figure 4 AMPK is not essential for Hippo pathway activation under energy stress.

(A–B) AMPK did not noticeably affect LATS1 and MST1 activation. SFB tagged LATS1 (A) or MST1 (B) was co-expressed with active AMPK truncation mutant in 293T cells. Cell lysates were subjected to western blotting with indicated antibodies. (C) LATS1 kinase activation was observed in both wild-type and AMPK-deficient cells. Wild-type MEFs (WT) and AMPKα-knockout MEFs (AMPKα KO) were treated with 2-DG (25 mM) in glucose-free medium and rapamycin (50 nM) for 4 hours. Western blotting analysis was performed using indicated antibodies. Uncropped images of western blots are shown in Supplementary Fig. 7.

Supplementary Figure 5 AMPK-regulated YAP phosphorylation does not affect 14-3-3 binding or subcellular localization of YAP, but suppresses its transcriptional activity.

(A) Phosphorylation of YAP by AMPK did not affect the association of YAP with the 14-3-3 protein. Indicated plasmids were co-expressed in 293T cells. Cell lysates were used for pulldown assay with S protein beads and subjected to western blotting. (B–C) Phosphorylation of YAP on the S61 site was not required for the binding of YAP to 14-3-3 (B). The active AMPK truncation mutant did not increase the association between the YAP-4SA-61S mutant and 14-3-3 (C). (D) AMPK did not affect cellular localisation of YAP. HeLa cells with exogenously expressed active AMPK truncation were subjected to immunostaining with the GST and YAP antibodies. Nuclei were visualized by Dapi. Scale bar = 20μm. (E) S61 mutation did not affect YAP localisation. HeLa cells with exogenously expressed indicated YAP mutants were subjected to immunostaining with Flag antibody. Nuclei were visualised by Dapi. Scale bar = 20μm. (F) Phospho-mimetic mutation of the S94 site (S94D), but not the 61 site (S61D), interfered with YAP-TEAD binding. Indicated plasmids were co-expressed in 293T cells. Cells lysates were used for pulldown assay with S protein beads and subjected to western blotting. (G) Phospho-mimetic mutants of the S61 site (S61D) and the S94 site (S94D) suppressed the transcription of YAP downstream genes. The transcripts of YAP-regulated genes (CTGF and CYR61) were detected by quantitative PCR in indicated YAP stable cells and normalised (mean ± s.d, n = 3 biological replicates). ∗∗∗P < 0.001(Student t-test). Statistics source data are shown in Supplementary Table 2. Uncropped images of western blots are shown in Supplementary Fig. 7.

Supplementary Figure 6 Predicted TEAD-binding site in the GLUT3 promoter region.

A conserved TEAD binding site was identified in the GLUT3 promoter region. ENCODE data showed that this site was occupied by TEAD transcription factor.

Supplementary Figure 7 A proposed model of YAP regulation by AMPK and Hippo pathway under energy stress.

YAP is phosphorylated and suppressed in response to energy stress. Both LATS kinase and AMPK are activated by energy stress and contribute to YAP inhibition via phosphorylating it on multiple sites. Energy stress induced-LATS kinase activation largely depends on the suppression of Rho GTPase and actin cytoskeleton dynamics, but it is also indirectly regulated by AMPK. With glucose is abundant, YAP translocates into nucleus and promotes glycolysis at least in part by upregulating GLUT3 at transcriptional level.

Supplementary Table 1 Identification of YAP phosphorylation site by AMPK

Supplementary information

Supplementary Information

Supplementary Information (PDF 2049 kb)

Supplementary Table 1

Supplementary Information (XLSX 35 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, W., Xiao, ZD., Li, X. et al. AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nat Cell Biol 17, 490–499 (2015). https://doi.org/10.1038/ncb3113

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3113

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing